Small Molecule
C30H25F10NO3
875446-37-0
Anacetrapib represents a significant paradox in modern cardiovascular drug development. As a potent inhibitor of cholesteryl ester transfer protein (CETP), it achieved unprecedented success in modifying lipid profiles, most notably by more than doubling high-density lipoprotein cholesterol (HDL-C). In the landmark REVEAL trial, it became the only drug in its class to demonstrate a statistically significant, albeit modest, reduction in major coronary events. However, this clinical success was overshadowed by a complex profile: the therapeutic benefit was attributed not to its primary HDL-raising mechanism but to a secondary reduction in non-HDL cholesterol. This, combined with its unique and concerning pharmacokinetic property of long-term accumulation in adipose tissue, created a challenging risk-benefit and commercial proposition. Ultimately, in October 2017, its developer, Merck & Co., abandoned the program, closing a tumultuous chapter for the CETP inhibitor hypothesis. This report provides an exhaustive analysis of Anacetrapib's journey, dissecting its scientific basis, clinical trial evidence, and the strategic factors that led to its discontinuation, offering critical lessons for the future of lipid-modifying therapies.
Anacetrapib is a small molecule drug developed as an orally administered agent for the treatment of dyslipidemia.[1] It is identified by the Chemical Abstracts Service (CAS) Number 875446-37-0 and the DrugBank accession number DB06630.[1] During its development, it was also referred to by the code MK-0859 and the non-proprietary name anacetrapibum.[1]
The molecular formula of Anacetrapib is C30H25F10NO3, corresponding to a molar mass of approximately 637.51 g·mol⁻¹.[2] Its formal International Union of Pure and Applied Chemistry (IUPAC) name is (4S,5R)-5-[3,5-bis(trifluoromethyl)phenyl]-3-[[2-(4-fluoro-2-methoxy-5-propan-2-ylphenyl)-5-(trifluoromethyl)phenyl]methyl]-4-methyl-1,3-oxazolidin-2-one.[1] This complex, highly fluorinated chemical structure is a defining characteristic of the molecule and is fundamental to understanding its biological behavior. The presence of ten fluorine atoms, primarily within three trifluoromethyl (
CF3) groups, imparts a high degree of lipophilicity (fat-solubility) to the compound.[1] This intrinsic chemical property has profound downstream consequences for its pharmacokinetics, directly influencing its poor aqueous solubility, its absorption characteristics, and, most critically, its extensive distribution into and long-term retention within adipose tissue.[3] The connection between this molecular structure and the drug's ultimate clinical and regulatory challenges is a central theme in its developmental history.
A consolidation of its key chemical identifiers is provided in Table 1 for reference.
Identifier | Value | Source(s) |
---|---|---|
Common Name | Anacetrapib | 2 |
DrugBank ID | DB06630 | 1 |
CAS Number | 875446-37-0 | 1 |
Type | Small Molecule | 1 |
Molecular Formula | C30H25F10NO3 | 2 |
Molar Mass | 637.51 g·mol⁻¹ | 2 |
IUPAC Name | (4S,5R)-5-[3,5-bis(trifluoromethyl)phenyl]-3-[[2-(4-fluoro-2-methoxy-5-propan-2-ylphenyl)-5-(trifluoromethyl)phenyl]methyl]-4-methyl-1,3-oxazolidin-2-one | 1 |
UNII | P7T269PR6S | 1 |
ChEMBL ID | CHEMBL1800807 | 1 |
Anacetrapib is pharmacologically classified as a lipid-modifying agent, and more specifically, as an anticholesteremic agent.[1] Its mechanism of action places it in the therapeutic class of cholesteryl ester transfer protein (CETP) inhibitors.[1] Chemically, it is a derivative of the oxazolidinone class.[7]
The development of Anacetrapib and the entire CETP inhibitor class was predicated on the long-standing "HDL hypothesis".[9] This hypothesis emerged from decades of epidemiological research showing a strong, consistent, and inverse correlation between levels of high-density lipoprotein cholesterol (HDL-C, often termed "good cholesterol") and the risk of atherosclerotic cardiovascular disease.[10] Further support came from genetic studies of individuals with CETP deficiency, who exhibit markedly elevated HDL-C levels and a correspondingly lower risk of coronary heart disease.[10] The biological role of CETP is to facilitate the transfer of cholesteryl esters from HDL particles to atherogenic, apolipoprotein B-containing lipoproteins such as very-low-density lipoprotein (VLDL) and low-density lipoprotein (LDL).[6] Therefore, the pharmacological inhibition of CETP presented a logical and highly attractive therapeutic strategy to raise HDL-C levels and, theoretically, reduce cardiovascular risk.[15] Anacetrapib was developed by Merck & Co. as a potent and selective agent to test this very premise in a large-scale clinical setting.[2] This scientific context is essential for appreciating the profound impact of its eventual clinical trial results, which ultimately challenged the foundational hypothesis upon which the drug was built.
The primary mechanism of action of Anacetrapib is the potent and reversible inhibition of the plasma glycoprotein CETP.[1] CETP plays a central role in reverse cholesterol transport by mediating the heteroexchange of neutral lipids between lipoproteins. Specifically, it transfers atheroprotective cholesteryl esters (CE) from the core of HDL particles to pro-atherogenic, apolipoprotein B-containing lipoproteins (VLDL and LDL) in exchange for triglycerides (TGs).[6]
By binding to and inhibiting CETP, Anacetrapib effectively blocks this transfer process.[6] This blockade has two major consequences on HDL metabolism. First, it prevents the depletion of CE from HDL particles, leading to the accumulation of larger, CE-enriched HDL particles (specifically, the HDL2 subfraction).[13] Second, these larger HDL particles are catabolized and cleared from the circulation more slowly than their smaller counterparts, further contributing to a net increase in the plasma concentration of HDL-C and its primary structural protein, apolipoprotein A-I (ApoA-I).[10]
The high potency of Anacetrapib was a critical feature that distinguished it from earlier CETP inhibitors and was likely a prerequisite for its clinical efficacy. The history of the CETP inhibitor class is marked by a series of high-profile failures. Torcetrapib failed due to significant off-target toxicities, including elevations in blood pressure and aldosterone.[2] The subsequent agent, dalcetrapib, was found to be safe but was a much weaker inhibitor, producing only a modest increase in HDL-C of about 30% with no effect on LDL-C; it was ultimately abandoned for lack of efficacy.[2] Anacetrapib, by contrast, was a highly potent inhibitor capable of producing dramatic elevations in HDL-C (>100%) and, importantly, significant reductions in LDL-C.[21] This suggests a clear potency-response relationship within the class, where the modest effects of dalcetrapib were simply insufficient to alter the overall atherogenic lipid profile in a clinically meaningful way. Anacetrapib's potency allowed it to have a much broader and more profound impact on lipoprotein metabolism, particularly on the atherogenic non-HDL particles, which was ultimately the source of its clinical benefit in the REVEAL trial.
The pharmacodynamic effect of Anacetrapib on circulating lipoproteins is profound and multifaceted. Its most prominent effect, consistent with its primary mechanism, is a substantial increase in HDL-C levels. Across multiple clinical trials, Anacetrapib consistently produced mean increases in HDL-C ranging from 102% to as high as 139% over placebo.[7] This was accompanied by a significant increase in ApoA-I, the major protein component of HDL, by 36% to 45%.[13]
In addition to its effects on HDL, Anacetrapib also exerts a significant lowering effect on pro-atherogenic lipoproteins. In early- and mid-stage trials, it demonstrated LDL-C reductions of approximately 40%.[21] In the large-scale REVEAL trial, where LDL-C was measured by the more accurate method of beta-quantification (ultracentrifugation) in a subset of patients, the reduction was a more modest but still significant 17% from an already low baseline.[10] This was reflected in an 18% reduction in non-HDL cholesterol (a measure of all atherogenic lipoproteins) and an 18% to 21% reduction in apolipoprotein B (ApoB), which represents the total number of atherogenic particles.[13]
Furthermore, Anacetrapib demonstrated a consistent and potentially important ancillary benefit: the reduction of lipoprotein(a) [Lp(a)]. Lp(a) is a highly atherogenic lipoprotein particle whose levels are genetically determined and are not significantly lowered by statin therapy.[14] Across trials, Anacetrapib reduced Lp(a) levels by 25% to 36%.[13] While the clinical benefit observed in the REVEAL trial was largely attributed to the reduction in non-HDL-C, the concurrent lowering of a known independent and causal cardiovascular risk factor like Lp(a) may have contributed to the overall risk reduction. This effect represents a potentially valuable mechanism that distinguishes Anacetrapib from traditional statin therapy and suggests that its clinical profile may be more complex than a simple non-HDL-C lowering effect. This reframes the potential value of future agents in this class, shifting focus away from the discredited HDL hypothesis and toward the simultaneous targeting of multiple distinct atherogenic lipoproteins.
The robust LDL-C lowering effect of Anacetrapib, which was more pronounced than that seen with other CETP inhibitors, prompted investigation into mechanisms beyond direct CETP inhibition. A critical discovery was that Anacetrapib has a CETP-independent effect on the proprotein convertase subtilisin/kexin type 9 (PCSK9) pathway.[13] PCSK9 is a circulating protein that binds to the LDL receptor (LDLR) on the surface of liver cells, targeting it for lysosomal degradation. By promoting LDLR degradation, PCSK9 reduces the liver's capacity to clear LDL particles from the blood, thereby raising plasma LDL-C levels.[13]
Studies demonstrated that Anacetrapib reduces plasma concentrations of PCSK9.[13] This reduction in PCSK9 levels leads to a decrease in LDLR degradation, resulting in a higher density of LDL receptors on the hepatocyte surface. This, in turn, enhances the clearance of LDL particles and VLDL remnants from the circulation, providing a clear biological mechanism for the observed reduction in LDL-C and ApoB.[13] This finding was pivotal because it provided a validated, non-HDL-related explanation for a significant portion of Anacetrapib's efficacy. It helped to shift the scientific narrative away from the increasingly dubious HDL hypothesis and toward the well-established paradigm of LDL-lowering. By acting, in part, as a PCSK9-lowering agent, Anacetrapib's effects became more understandable through the familiar lens of LDLR biology. This allowed the modest clinical benefit seen in the REVEAL trial to be interpreted as a predictable consequence of its non-HDL-C lowering effect, lending credibility to the result while simultaneously undermining the drug's original therapeutic rationale.[13]
The mechanistic picture is complex, as some in vitro studies in cultured liver cells unexpectedly showed that Anacetrapib could also directly downregulate the expression of both LDLR and PCSK9 at the transcriptional level, an effect mediated through the sterol regulatory element-binding protein (SREBP) pathway.[26] This suggests a more intricate regulatory role, but the net
in vivo effect observed in humans was a clear reduction in plasma PCSK9 and an enhancement of LDL particle catabolism.[13] Other proposed ancillary mechanisms for LDL-C reduction include compositional changes to LDL particles (e.g., increased triglyceride content) that increase their affinity for the LDLR, and reduced expression of the inducible degrader of the LDLR (IDOL).[13]
The superior potency and broader inhibitory profile of Anacetrapib compared to its predecessors can be traced to its specific molecular interactions with the CETP enzyme. Atomistic molecular dynamics simulations have provided detailed insights into this drug-protein binding.[15] These studies show that Anacetrapib has a strong affinity for the concave surface of the CETP molecule, which is the surface that interacts with lipoproteins.[15]
The primary binding site for Anacetrapib is proposed to be within the long, hydrophobic tunnel that traverses the core of the CETP molecule. This tunnel serves as the conduit for the passage of neutral lipids like CE and TGs.[15] By lodging itself within this tunnel, particularly near the N-terminal opening, Anacetrapib acts as a physical plug, sterically hindering the diffusion of CE out of the protein and thereby directly inhibiting its function.[15] Furthermore, simulations suggest Anacetrapib can allosterically regulate the conformation of helix X, a flexible structural region of CETP that is critical for the process of lipid exchange with lipoproteins.[15]
This binding mode contrasts sharply with that of the less potent inhibitor, dalcetrapib. Biochemical studies have shown that Anacetrapib and dalcetrapib bind to different sites on the CETP molecule and have distinct effects on lipid transfer between HDL subfractions; for instance, Anacetrapib inhibits the transfer of CE from smaller HDL3 to larger HDL2 particles, whereas dalcetrapib does not.[28] This difference in binding site—with Anacetrapib acting as a direct tunnel blocker and dalcetrapib inducing a less effective conformational change from a different site—provides a clear molecular basis for Anacetrapib's greater pharmacodynamic potency. This demonstrates that "CETP inhibition" is not a monolithic mechanism; rather, the precise nature of the drug-protein interaction dictates the extent and character of the resulting lipid modulation. Anacetrapib's more effective binding mode translated directly into a more profound lipid-modifying effect, which was a necessary condition for its eventual, albeit modest, clinical success.
The pharmacokinetic profile of Anacetrapib is highly unusual and is central to understanding both its clinical effects and the ultimate decision to halt its development. It is characterized by a pronounced food effect, extensive tissue distribution, and an exceptionally long terminal half-life driven by sequestration in adipose tissue. A summary of its key pharmacokinetic parameters is presented in Table 2.
Following oral administration, Anacetrapib is rapidly absorbed, with the time to reach maximum plasma concentration (Tmax) occurring at approximately 4 hours post-dose in both fasted and fed states.[5] However, the extent of its absorption is profoundly influenced by the presence of food, a direct consequence of its high lipophilicity and poor aqueous solubility. Administration with a low-fat meal can increase drug exposure (as measured by area under the curve [AUC] and maximum concentration [
Cmax]) by two- to three-fold compared to the fasted state.[5] This effect is even more dramatic with a high-fat meal, which can increase exposure by as much as six- to eight-fold.[5] Studies have also shown that plasma exposure tends to increase in a less than dose-proportional manner, particularly in the fasted state, suggesting that absorption becomes saturated at higher doses.[5]
The most defining and problematic feature of Anacetrapib's pharmacokinetics is its extensive distribution and accumulation in adipose tissue. Its highly lipophilic chemical structure gives it a strong affinity for fatty tissues, causing the drug to partition out of the plasma and into fat stores throughout the body.[4] This leads to the formation of a massive, long-term drug reservoir.[30]
Preclinical studies in mice demonstrated this effect vividly, showing that while blood concentrations accumulated 3- to 9-fold during a 42-day dosing period, concentrations in white adipose tissue accumulated 20- to 40-fold.[4] This phenomenon was confirmed in human studies. A Phase I study involving adipose tissue biopsies revealed that Anacetrapib continued to accumulate in fat throughout the treatment period, even after plasma concentrations had reached a steady-state plateau.[30] This accumulation in adipose tissue was a well-documented finding throughout its clinical development and was explicitly noted as a safety concern during the REVEAL trial.[6]
Anacetrapib is primarily metabolized in the liver by the cytochrome P450 enzyme CYP3A4.[16] The main metabolic pathways involve O-demethylation and hydroxylation at the biphenyl and isopropyl moieties, resulting in three main oxidative metabolites (M1, M2, and M3) that are considered to be nearly inactive.[16]
Following metabolism, the drug is eliminated almost exclusively via the feces, with the majority recovered as the unchanged parent compound. Renal excretion is negligible, with less than 0.1% of an administered dose being recovered in the urine.[16] This excretion profile suggests that renal impairment is unlikely to be a major limitation for its use. However, because it is a moderately sensitive substrate of CYP3A4, conditions that inhibit this enzyme, such as severe hepatic impairment or co-administration of potent CYP3A4 inhibitors, could lead to a significant increase in Anacetrapib plasma levels.[37]
The extensive sequestration of Anacetrapib in adipose tissue results in a biphasic elimination profile and an unprecedentedly long terminal elimination half-life.[16] After a single dose, the apparent terminal half-life ranges from approximately 9 to 83 hours, depending on the fed state.[5] With multiple daily doses, a steady state in plasma is reached after about 7 days, with an effective half-life of approximately 20 hours governing the fluctuations between doses.[16]
However, this is overshadowed by a much longer terminal elimination phase, which becomes increasingly apparent with longer treatment durations. This phase is driven by the very slow release of the drug from the deep adipose tissue compartment back into the circulation. The clinical consequence is that Anacetrapib persists in the body for years after the last dose is taken. In a follow-up to the DEFINE trial, low but detectable plasma concentrations of the drug were measured up to four years after treatment discontinuation.[16] Pharmacokinetic modeling estimated the half-life of elimination from this third (adipose) compartment to be approximately 550 days.[16]
This uniquely long residence time created an intractable safety and regulatory problem. Standard principles of drug development require a predictable pharmacokinetic profile that allows for the drug to be cleared from the body in a reasonable timeframe should a serious adverse effect emerge. With Anacetrapib, if a rare but severe long-term toxicity (e.g., a specific malignancy or neurodegenerative disorder) were to be discovered after years of market use, patients would remain exposed to the drug for years after its withdrawal, with no practical way to accelerate its elimination. This potential for an unknown, delayed, and essentially irreversible toxicity represented a major, and perhaps insurmountable, safety liability. While the REVEAL trial and its long-term follow-up did not identify such a hazard, the risk of one emerging in the future weighed heavily against the drug's overall benefit-risk profile and was a critical factor in the decision to abandon its development.[6]
Parameter | Value | Source(s) |
---|---|---|
Tmax | ~4 hours | 5 |
Absorption Food Effect (High-Fat Meal) | ~6- to 8-fold increase in exposure | 5 |
Primary Metabolism | CYP3A4 | 16 |
Primary Excretion Route | Feces | 16 |
Apparent Terminal Half-life (single dose) | 42–83 hours (fed) | 37 |
Effective Half-life (multiple dose) | ~20 hours | 37 |
Key Distribution Feature | Extensive accumulation in adipose tissue | 4 |
Half-life from Adipose Compartment | ~550 days | 16 |
The clinical development of Anacetrapib cannot be understood in isolation; it was profoundly shaped by the repeated and dramatic failures of other drugs in its class. This history created a climate of intense skepticism and set an exceptionally high bar for success. A comparative summary of the four major late-stage CETP inhibitors is presented in Table 3.
The first agent to reach large-scale outcomes testing was torcetrapib, developed by Pfizer. Its development was abruptly terminated in December 2006 when the Phase III ILLUMINATE trial was stopped early due to a clear signal of harm, including an excess of cardiovascular events and all-cause mortality in the treatment arm.[2] Subsequent analyses attributed this adverse outcome to off-target effects of the molecule, unrelated to CETP inhibition, which caused increases in blood pressure, sodium retention, and aldosterone levels.[19]
The second agent was dalcetrapib, developed by Hoffmann-La Roche. While it did not exhibit the off-target toxicities of torcetrapib, it was a much less potent CETP inhibitor. In May 2012, its development was also halted after the dal-OUTCOMES trial was stopped for futility, showing no clinical benefit.[2] Its failure was largely attributed to its weak pharmacodynamic effect, as it produced only a modest increase in HDL-C with no significant effect on LDL-C.[10]
The third major failure was evacetrapib, developed by Eli Lilly. This agent was potent, producing lipid changes comparable to Anacetrapib, and appeared to be free of torcetrapib's off-target liabilities. Despite this promising profile, its development was terminated in October 2015 when its large Phase III ACCELERATE trial was stopped for futility, again demonstrating no reduction in cardiovascular events.[2]
The successive failures of these three agents created a formidable challenge for Anacetrapib. The torcetrapib failure could be rationalized as drug-specific toxicity, and the dalcetrapib failure as a lack of potency. However, the failure of the potent and seemingly "clean" evacetrapib was a major blow to the entire therapeutic hypothesis, suggesting that even ideal CETP inhibition might not be clinically effective. This history placed immense pressure on the REVEAL trial, framing it not merely as a test of Anacetrapib, but as the final, definitive referendum on the viability of the entire CETP inhibitor class. When Merck announced that REVEAL would continue after the evacetrapib failure, it was a significant strategic decision, betting that their trial's larger size and longer duration would succeed where others had failed.[20]
Drug (Developer) | Key Trial | HDL-C Effect | LDL-C Effect | Key Safety/Efficacy Finding | Development Status |
---|---|---|---|---|---|
Torcetrapib (Pfizer) | ILLUMINATE | +72% | -25% | Increased mortality and CV events (off-target toxicity) | Terminated 2006 |
Dalcetrapib (Roche) | dal-OUTCOMES | +30–40% | No effect | Lack of efficacy | Terminated 2012 |
Evacetrapib (Eli Lilly) | ACCELERATE | +129–133% | -36% | Lack of efficacy | Terminated 2015 |
Anacetrapib (Merck) | REVEAL | +104–139% | -17% to -40% | Modest reduction in coronary events, but development halted | Terminated 2017 |
[2]
The early clinical development of Anacetrapib focused on establishing its pharmacokinetic (PK) and pharmacodynamic (PD) profile and, most importantly, demonstrating that it was free from the adverse effects that plagued torcetrapib. Phase I studies in healthy volunteers successfully characterized its absorption, including the significant food effect, and confirmed its potent, dose-dependent inhibition of CETP activity.[29] These initial human studies were crucial in showing that Anacetrapib was generally well-tolerated and, unlike torcetrapib, did not cause adverse changes in blood pressure, electrolytes, or aldosterone levels.[11] A dedicated Phase I study in healthy Chinese subjects later confirmed that its PK profile was comparable across different ethnic populations.[40]
Phase II studies expanded this evaluation into patient populations. These included a dose-ranging study (NCT00325455) in patients with hypercholesterolemia or mixed dyslipidemia, which was terminated, and a study evaluating Anacetrapib as both monotherapy and in combination with atorvastatin in Japanese patients with dyslipidemia.[41] These studies consistently demonstrated the drug's robust lipid-modifying effects and continued to support its favorable safety profile.[42]
The pivotal mid-stage trial for Anacetrapib was the DEFINE (Determining the EFficacy and Tolerability of CETP INhibition with AnacEtrapib) study. This was a 76-week, randomized, double-blind, placebo-controlled trial that enrolled 1,623 patients with established coronary heart disease (CHD) or a CHD risk equivalent who were already on stable statin therapy.[8]
The DEFINE trial had two primary objectives: to quantify the lipid-modifying efficacy of Anacetrapib and to rigorously assess its safety profile over an extended period. The results, reported in 2010, were highly successful on both fronts. In terms of efficacy, Anacetrapib 100 mg daily produced dramatic changes in lipoproteins compared to placebo: HDL-C increased by 138.1%, and LDL-C decreased by 39.8%.[19]
From a strategic perspective, the safety results were even more critical. The trial confirmed the findings from Phase I studies, showing an acceptable side-effect profile with no significant differences between the Anacetrapib and placebo groups in blood pressure, serum electrolytes, or aldosterone levels.[8] Furthermore, a prospective adjudication of cardiovascular events found a numerically lower rate in the Anacetrapib group (2.0%) compared to the placebo group (2.6%), providing strong evidence that Anacetrapib did not share the adverse cardiovascular effects of torcetrapib.[44]
The DEFINE trial was the crucial de-risking step in the Anacetrapib development program. By demonstrating that potent CETP inhibition could be achieved safely, without the specific off-target toxicities that doomed torcetrapib, it provided Merck with the necessary confidence to proceed with the massive, high-risk, and costly REVEAL cardiovascular outcomes trial. The clean safety and robust efficacy profile from DEFINE was the essential green light that kept the program—and the hopes for the CETP inhibitor class—alive.
The REVEAL (Randomised EValuation of the Effects of Anacetrapib through Lipid-modification) trial, also known by its protocol identifier HPS3/TIMI55, was the definitive cardiovascular outcomes study for Anacetrapib.[22] It was a large-scale, randomized, double-blind, placebo-controlled trial designed with the statistical power and duration necessary to robustly assess the clinical efficacy and safety of the drug.[22]
The trial enrolled 30,449 adult participants with pre-existing atherosclerotic vascular disease, including a history of myocardial infarction, cerebrovascular disease, or peripheral arterial disease.[22] Participants were recruited from over 430 centers across North America, Europe, and China.[45] A key design feature was that all participants were treated with intensive atorvastatin therapy to achieve effective control of LDL-C prior to and during the trial, with a mean baseline LDL-C of just 61 mg/dL.[22] Patients were then randomized to receive either Anacetrapib 100 mg once daily or a matching placebo, added to their statin regimen.[22] The median follow-up period was 4.1 years.[22] The study was designed and coordinated by independent academic investigators from the Clinical Trial Service Unit (CTSU) at the University of Oxford and the TIMI Study Group, with funding provided by Merck.[23]
The primary efficacy outcome of the REVEAL trial was the time to the first major coronary event (MCE), defined as a composite of coronary death, myocardial infarction (MI), or coronary revascularization.[22]
The trial successfully met its primary endpoint. Treatment with Anacetrapib resulted in a statistically significant 9% proportional reduction in the risk of MCE. The event occurred in 10.8% of patients in the Anacetrapib group compared to 11.8% of patients in the placebo group, yielding a rate ratio (RR) of 0.91 (95% Confidence Interval [CI], 0.85 to 0.97; p=0.004).[22]
Analysis of the secondary outcomes provided further nuance. The risk of MI was significantly reduced in the Anacetrapib group (4.4% vs. 5.1%; p=0.007).[48] However, there was no significant reduction in the risk of coronary death (2.5% vs. 2.8%;
p=0.25).[48] Another key secondary endpoint, a composite of major atherosclerotic events (MCE plus presumed ischemic stroke), did not reach statistical significance (RR 0.93; 95% CI, 0.86 to 1.00;
p=0.052), a result attributed to a lack of observed benefit on stroke.[25] A summary of these key outcomes is presented in Table 4.
Outcome | Anacetrapib Group (%) | Placebo Group (%) | Rate Ratio (95% CI) | P-value |
---|---|---|---|---|
Primary Endpoint (Coronary Death, MI, or Revascularization) | 10.8 | 11.8 | 0.91 (0.85–0.97) | 0.004 |
Myocardial Infarction (MI) | 4.4 | 5.1 | N/A | 0.007 |
Coronary Death | 2.5 | 2.8 | N/A | 0.25 |
Major Atherosclerotic Events (Primary + Stroke) | N/A | N/A | 0.93 (0.86–1.00) | 0.052 |
New-Onset Diabetes | 5.3 | 6.0 | N/A | 0.05 |
[22]
While the REVEAL trial was a statistical success, its interpretation proved to be a major turning point for the field of lipidology. At the trial's midpoint, Anacetrapib had produced the expected lipid changes: HDL-C was increased by a massive 104%, while non-HDL-C was reduced by a more modest 18%.[10]
The central question was whether the observed 9% clinical benefit was driven by the unprecedented rise in HDL-C or the modest fall in non-HDL-C. The overwhelming consensus among the trial investigators and external experts was that the benefit was largely, if not entirely, attributable to the reduction in atherogenic non-HDL-C particles.[10] The magnitude of the risk reduction was consistent with what would have been predicted from an 18% non-HDL-C lowering based on extensive meta-analyses of statin trials.[13] If the HDL hypothesis were correct, a >100% increase in HDL-C should have produced a far greater clinical benefit. The fact that it did not strongly implied that the HDL particles generated through CETP inhibition were either not functionally anti-atherogenic, or that their protective effect was negligible compared to the detrimental effect of ApoB-containing lipoproteins.
Thus, the REVEAL trial, designed as the ultimate test of the HDL hypothesis, ironically became one of the final pieces of evidence arguing against it as a primary therapeutic target. It demonstrated that even a profound, pharmacologically-induced increase in HDL-C does not confer clinical benefit beyond that which is achieved by lowering atherogenic lipoproteins. This finding helped to decisively shift the focus of cardiovascular drug development away from raising HDL-C and reinforced the primacy of lowering LDL-C, non-HDL-C, and ApoB as the central goal of lipid-modifying therapy.
Following the completion of the 4.1-year treatment period, surviving participants were enrolled in a post-trial follow-up phase for a median of an additional 2.2 years, during which they were no longer taking the study drug.[45] The results of this extended follow-up, published in 2021, provided further critical insights.
Remarkably, the clinical benefit of Anacetrapib appeared to grow over time. During the post-trial period, there was a further 20% reduction in major coronary events in the group originally allocated to Anacetrapib.[51] Over the entire 6.3-year observation period (in-trial plus post-trial), the total proportional risk reduction was 12% (
p<0.001).[51] This highly unusual "legacy effect" is most logically explained by the drug's unique pharmacokinetics. The slow washout of Anacetrapib from the massive adipose tissue reservoir likely resulted in continued, low-level drug exposure and sustained lipid-modifying effects for years after treatment cessation.[16] This finding underscores the importance of both the magnitude and the
duration of lipid-lowering in reducing cardiovascular risk and suggests that the 4.1-year result from the main trial may have actually underestimated the drug's true long-term efficacy.
Crucially, from a safety perspective, no new adverse signals emerged during the extended follow-up. There was no evidence of any late-emerging harm related to non-vascular mortality, cancer, or other serious adverse events, providing some reassurance regarding the long-term consequences of its tissue accumulation.[45]
Across its entire clinical development program, from early phase studies to the large-scale REVEAL trial, Anacetrapib was generally well-tolerated.[7] In the definitive REVEAL trial, there were no statistically significant differences between the Anacetrapib and placebo groups in the rates of overall serious adverse events, drug-related adverse events leading to discontinuation, cause-specific mortality, or the incidence of cancer.[18] This favorable general safety profile was a key feature that distinguished it from the first-generation CETP inhibitor, torcetrapib.
Despite its overall good tolerability, detailed analysis from the REVEAL trial identified several small but statistically significant physiological effects.
The most significant safety concern surrounding Anacetrapib was not an observed adverse event, but rather a theoretical long-term risk stemming from its unique pharmacokinetics. The extensive and persistent accumulation of the drug in adipose tissue was a consistent finding throughout its development.[4] This high lipophilicity and slow elimination created a drug reservoir that would expose patients to the compound for years after they stopped taking it.[31]
This created a difficult toxicological question: what are the potential consequences of very long-term, low-level exposure to a lipophilic compound stored in fat cells? While preclinical studies in mice were reassuring—showing that high levels of accumulated Anacetrapib did not appear to impair adipose tissue function and that the drug was not suddenly mobilized from fat stores during rapid weight loss—these findings could not definitively rule out a long-latency adverse effect in humans.[54] The extended follow-up from REVEAL also provided reassurance up to a median of 6.3 years, but could not address risks that might emerge after 10, 15, or 20 years. This unquantifiable, long-term potential for an unknown and irreversible toxicity became a central element of the drug's risk profile and a major consideration in the decision to halt its development.[6]
A critical achievement of the Anacetrapib development program was to successfully uncouple potent CETP inhibition from the specific off-target toxicities that doomed torcetrapib. Rigorous evaluation in both early-phase studies and the large DEFINE and REVEAL trials confirmed that Anacetrapib did not cause the clinically significant increases in blood pressure, aldosterone stimulation, or adverse electrolyte disturbances (e.g., hypokalemia) that were the downfall of torcetrapib.[11] This demonstrated that those adverse effects were specific to the torcetrapib molecule and not an inherent class effect of CETP inhibition.
However, this created a trade-off in the nature of the safety risk. The risk associated with torcetrapib was acute, observable during treatment, and related to known physiological pathways. In contrast, Anacetrapib's safety profile was characterized by the introduction of a novel, chronic, post-treatment risk that was unpredictable and potentially irreversible due to its pharmacokinetic properties. This shifted the safety debate from managing known side effects to contending with a long-term, unquantifiable liability, fundamentally altering the drug's overall risk-benefit assessment.
Despite achieving a statistically significant positive result in its landmark cardiovascular outcomes trial—a first for the CETP inhibitor class—the development of Anacetrapib came to an end. On October 11, 2017, Merck & Co. announced that, following a thorough review of the complete clinical profile and discussions with external experts, the company would not be submitting applications for regulatory approval to the FDA or other global health authorities.[55] In the official statement, Dr. Roger M. Perlmutter, then-president of Merck Research Laboratories, concluded that "the clinical profile for anacetrapib does not support regulatory filings".[55]
Merck's decision was not based on a single flaw but on a comprehensive evaluation of the drug's suboptimal profile across three key domains: clinical efficacy, long-term safety, and commercial viability.
Ultimately, Anacetrapib was a drug that was statistically successful but clinically and commercially unviable. The combination of a modest, non-novel benefit, a unique and concerning long-term safety liability, and a difficult market position made the risk and expense of a global launch unjustifiable.
Despite its discontinuation, the Anacetrapib program yielded invaluable scientific knowledge that has had a lasting impact on the field.
Anacetrapib will be remembered as the most advanced and clinically successful of the CETP inhibitors, a class of drugs born from the compelling but ultimately flawed HDL hypothesis. Its development journey, culminating in the REVEAL trial, provided invaluable scientific insights, most notably by reinforcing the primacy of lowering atherogenic lipoproteins over raising HDL-C. While the drug demonstrated a modest but statistically significant cardiovascular benefit, its unique pharmacokinetic profile—characterized by profound and persistent accumulation in adipose tissue—created an untenable long-term safety risk. This, combined with the modest magnitude of its clinical efficacy and a challenging commercial landscape, led Merck & Co. to make the rational decision to halt its development. The story of Anacetrapib serves as a powerful case study in the complexities of drug development, illustrating that statistical success in a clinical trial is only one component of a much larger equation that includes clinical meaningfulness, long-term safety, and strategic market fit. Its legacy is not as a failed drug, but as a critical scientific tool that helped refine our understanding of lipoprotein metabolism and cardiovascular risk, paving the way for future therapeutic innovations.
Published at: September 19, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.